Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
F S Sci ; 4(2): 151-162, 2023 05.
Article in English | MEDLINE | ID: mdl-37011812

ABSTRACT

OBJECTIVE: To gain an understanding of the potential role of endoplasmic reticulum (ER) stress in the endometrial compartment during early pregnancy, a highly understudied area. DESIGN: This study examined the regulation of interferon-ß (IFNß) in response to ER stress in human decidualized and nondecidualized endometrial cells (human endometrial stromal cells [HESCs]) in vitro. In vivo, we examined ER stress and the IFNß levels locally in the mouse endometrium before and after implantation at embryonic day (E)1, E3, and E6. SETTING: The study was performed in a reproductive sciences laboratory for Human Growth and Development. PATIENT(S): None. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Quantitative polymerase chain reaction, Western blotting, and immunohistochemical analysis allowed us to test the action of endogenous ER stress activation in the endometrial compartment likely triggered by implantation and its ability to increase the endometrial IFNß levels. RESULT(S): In vitro, we observed a significant difference in the IFNß levels in HESCs, in response to ER stress activation, where decidualized HESCs exhibited a threefold increase in the IFNß levels compared with nondecidualized HESCs. Apoptotic caspase-3 activation was also isolated to the decidualized cells as a result of ER stress-dependent suppression of nuclear factor-kappa beta-regulated antiapoptotic factors, XIAP and MCL-1. In vivo, mouse endometrial IFNß was present in F4/80-positive macrophages at all time points examined. After implantation (E6), the mouse luminal epithelial cells robustly coexpressed both IFNß and the ER stress marker immunoglobulin heavy chain binding protein (BiP). CONCLUSION(S): These analyses demonstrate that both in vivo and in vitro, differentiated and decidualized endometrial cells undergoing ER stress have the capacity to produce increased IFNß levels; therefore, ER stress activation in the endometrial compartment may play a vital role in promoting successful implantation events.


Subject(s)
Embryo Implantation , Endometrium , Pregnancy , Female , Humans , Animals , Mice , Embryo Implantation/physiology , Cell Differentiation , Interferon-beta/metabolism
2.
F S Sci ; 4(2): 141-150, 2023 05.
Article in English | MEDLINE | ID: mdl-36603702

ABSTRACT

OBJECTIVE: To examine the activation and consequence of uterine apoptotic caspase-3 action on 1 day after coitus (dpc) in the pregnant mouse. We have previously demonstrated that in a pregnant uterus, caspase-3 activation from mid to late gestation isolated to the myometrial compartment is largely nonapoptotic and controls uterine quiescence. Additionally, we had demonstrated that apoptotic caspase-3 activation isolated to the endometrial compartment at term regulated endometrial prostaglandin synthesis. DESIGN: Uteri were isolated from pseudopregnant and nonligated controls and unilateral and bilateral ligated uterine horn mouse models at 1, 3, and 6 dpc. Uteri were examined for apoptotic indices, such as caspase-3 activation and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling staining. Immunohistochemical analysis identified the site of uterine apoptotic caspase-3 activation. The truncated form of phospholipase A2 was examined as a measure of apoptotic caspase-3-mediated calcium independent phospholipase A2 (iPLA2) activation. RESULT(S): We identified the site and impact of uterine apoptotic caspase-3 activation using uteri isolated from nonpregnant control animals at estrous and diestrous and control pregnant mice at 1-19 dpc. Our analysis revealed that apoptotic caspase-3 and iPLA2 activation were limited to the endometrial compartments of the control and unilateral ligated uteri on 1 dpc and were not found in the pseudopregnant or bilateral ligated uterine horn or on 3 or 6 dpc in the control and unilateral ligated uteri. CONCLUSION(S): In this study, we determined that uterine caspase-3 activation on 1 dpc, which is endometrial and apoptotic in nature, may play a potential role in regulating the previously reported preimplantation surge in endometrial PGE2 synthesis through apoptotic caspase-3-mediated iPLA2 activation. Our data indicate that the presence of a conceptus on 1 dpc likely triggers an increase in endometrial apoptotic caspase-3-mediated iPLA2 activation. When activated, iPLA2 causes the hydrolysis of fatty acids, resulting in arachidonic acid release and PGE2 production, which has been demonstrated to act in a leutoprotective manner in early pregnancy, prolonging progesterone synthesis and promoting uterine receptivity.


Subject(s)
Dinoprostone , Uterus , Female , Pregnancy , Mice , Animals , Caspase 3 , Endometrium , Phospholipases A2
3.
Sci Rep ; 9(1): 4452, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872705

ABSTRACT

The elevated level of Steroidogenic Factor 1 (Nr5a1, Sf-1) expression in the male gonadal development pathway, post sex determination, implies a vital role in testis gonadal differentiation. In this study we generated Sertoli cell-specific Nr5a1 KO mice (SC-SF-1-/-) at E14.5, which coincides with testis development post sex determination, using the Amh-Cre mouse model. Analysis of SC-SF-1-/- (Sertoli cell specific Nr5a1 knockout) testes demonstrated apoptosis as early as E15. Further analysis revealed that SC-SF-1-/- gonads displayed lower MDM2 levels resulting in elevated TP53 levels, which we believe may lead to apoptosis of the Sertoli cell population, inferring the possibility that NR5A1 directly regulates MDM2 expression. By E15.5, the Sertoli cell and germ cell population declined in SC-SF-1-/- mice resulting in the disruption of seminiferous cords with limited cord structure remaining at E18.5. Due to the loss of Sertoli and germ cells, the testis weights of SC-SF-1-/- mice at 6-weeks were much reduced; however, SC-SF-1-/- seminal vesicles weights were comparable suggesting intact Leydig cell androgen production. We conclude that NR5A1 regulates the TP53 pathway during development, is essential for fetal Sertoli cell survival and controls the cell cycle of Sertoli cells during differentiation.


Subject(s)
Sertoli Cells/cytology , Steroidogenic Factor 1/metabolism , Testis/cytology , Testis/embryology , Animals , Anti-Mullerian Hormone/metabolism , Apoptosis/genetics , Cell Survival , Female , Gene Expression Regulation, Developmental , Integrases/genetics , Male , Mice, Knockout , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , SOX9 Transcription Factor/genetics , Sertoli Cells/physiology , Sex Determination Processes , Steroidogenic Factor 1/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
EBioMedicine ; 39: 520-530, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30502052

ABSTRACT

BACKGROUND: Circulating estrogen (E2) levels are high throughout pregnancy and increase towards term, however its local tissue specific actions vary across gestation. For example, myometrial E2 regulated uterotonic action is disabled until term, whereas it's proliferative function is maintained in the breast. We have identified gestationally regulated splicing events, mediated by hnRNPG and modulated by E2 that generate alternatively spliced estrogen receptor alpha (ERα) variants (ERΔ7 and ERα46) in the myometrium. These variants allow for differential, gestationally regulated, modulation of the uterotonic action of E2. METHODS: Human myometrium isolated from preterm and term non-laboring and laboring pregnant women were analyzed for ERα isoforms and splice factor levels. Lentiviral mediated shRNA knockdown of hnRNPG and overexpression of ERΔ7 were performed in human myometrial (hTERT-HM) cells. Functional 3D collagen contraction assays were executed. FINDINGS: ERΔ7 acts as a dominant negative repressor of the uterotonic action of ERα66 and ERα46 isoforms through the regulation of the myometrial gap junction protein GJA1. Elimination of hnRNPG inhibits the generation of ERΔ7 while overexpression of ERΔ7 inhibited GJA1 expression. Moreover in vivo human myometrial hnRNPG levels decline at term in an E2 dependent manner resulting in a withdrawal of ERΔ7 levels and its tocolytic action at term. INTERPRETATION: Our findings implicate the unique role of ERΔ7 as a modulator of myometrial quiescence and define the mechanism of ERΔ7 generation, through hormonally regulated splicing events. FUND: This study was supported by NIH OPRU U01 supplement (HD047905), University of Pittsburgh and Wayne State University Perinatal Research Initiative (USA).


Subject(s)
Connexin 43/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Myometrium/metabolism , Uterine Contraction/metabolism , Alternative Splicing , Cell Line , Estrogens/metabolism , Exons , Female , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Myometrium/cytology , Organ Specificity , Pregnancy , Protein Isoforms/metabolism , Uterine Contraction/genetics , Uterus/metabolism
5.
Cell Death Dis ; 9(10): 933, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224704

ABSTRACT

The prevention of apoptotic caspase 3 activation through biological preconditioning, mediated through the modulation of the unfolded protein response has been demonstrated to ameliorate multiple pathophysiologies. The maintenance of non-apoptotic caspase 3 activity by the unfolded protein response within the pregnant uterus has previously been proven to be critical in inhibiting uterine myocyte contractility during pregnancy. Here we report that the pregnant uterus utilizes an unfolded protein response-preconditioning paradigm to conserve myometrial caspase 3 in a non-apoptotic state in order to effectively inhibit uterine contractility thereby preventing the onset of preterm labor. In the absence of appropriate endogenous preconditioning during pregnancy, uterine caspase 3 is transformed from a non-apoptotic to an apoptotic phenotype. Apoptotic caspase 3 activation results in the precocious triggering of local uterine inflammatory signaling and prostaglandin production, consequently resulting in an increased incidence of preterm birth. These findings represent a paradigm shift in our understanding of how preconditioning promotes the maintenance of uterine non-apoptotic caspase 3 action during pregnancy preventing the onset of premature uterine contraction and therefore defining the timing of the onset of labor.


Subject(s)
Caspase 3/metabolism , Myometrium/cytology , Myometrium/metabolism , Unfolded Protein Response/physiology , Uterus/cytology , Uterus/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Caspase 3/genetics , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Pregnancy , Signal Transduction/genetics , Signal Transduction/physiology , Unfolded Protein Response/genetics
6.
FASEB J ; 31(9): 4037-4052, 2017 09.
Article in English | MEDLINE | ID: mdl-28559440

ABSTRACT

Preterm birth (PTB) is the leading cause of neonatal mortality and morbidity, with few prevention and treatment options. Uterine contraction is a central feature of PTB, so gaining new insights into the mechanisms of this contraction and consequently identifying novel targets for tocolytics are essential for more successful management of PTB. Here we report that myometrial cells from human and mouse express bitter taste receptors (TAS2Rs) and their canonical signaling components (i.e., G-protein gustducin and phospholipase C ß2). Bitter tastants can completely relax myometrium precontracted by different uterotonics. In isolated single mouse myometrial cells, a phenotypical bitter tastant (chloroquine, ChQ) reverses the rise in intracellular Ca2+ concentration ([Ca2+]i) and cell shortening induced by uterotonics, and this reversal effect is inhibited by pertussis toxin and by genetic deletion of α-gustducin. In human myometrial cells, knockdown of TAS2R14 but not TAS2R10 inhibits ChQ's reversal effect on an oxytocin-induced rise in [Ca2+]i Finally, ChQ prevents mouse PTBs induced by bacterial endotoxin LPS or progesterone receptor antagonist mifepristone more often than current commonly used tocolytics, and this prevention is largely lost in α-gustducin-knockout mice. Collectively, our results reveal that activation of the canonical TAS2R signaling system in myometrial cells produces profound relaxation of myometrium precontracted by a broad spectrum of contractile agonists, and that targeting TAS2Rs is an attractive approach to developing effective tocolytics for PTB management.-Zheng, K., Lu, P., Delpapa, E., Bellve, K., Deng, R., Condon, J. C., Fogarty, K., Lifshitz, L. M., Simas, T. A. M., Shi, F., ZhuGe, R. Bitter taste receptors as targets for tocolytics in preterm labor therapy.


Subject(s)
Gene Expression Regulation/physiology , Myometrium/cytology , Obstetric Labor, Premature/drug therapy , Receptors, G-Protein-Coupled/metabolism , Albuterol , Animals , Calcium/metabolism , Chloroquine , Female , Humans , Magnesium Sulfate , Mice , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Oxytocin/pharmacology , Phenanthrolines , Pregnancy , Quaternary Ammonium Compounds , Receptors, G-Protein-Coupled/genetics , Transducin/genetics , Transducin/metabolism
7.
J Cardiovasc Pharmacol Ther ; 22(4): 337-346, 2017 07.
Article in English | MEDLINE | ID: mdl-28376665

ABSTRACT

A broad definition of preconditioning is "the preparation for a subsequent action." Mounting evidence demonstrates that novel remote preconditioning paradigms, in which protective stimuli experienced locally can capacitate systemic tolerance and enhanced cell viability upon exposure to ensuing cellular insults, have been largely successful in the field of cardiovascular ischemia/reperfusion injury. To ensure successful protective preconditioning, some models (including the uterus) have been demonstrated to activate the unfolded protein response (UPR), which is a cellular stress response controlled at the level of the endoplasmic reticulum. However, in the context of remote preconditioning, activation of these intracellular molecular pathways must result in the extracellular transmission of adaptive signals to remote targets. In our recently published manuscript, we have described the activation of the UPR in the pregnant uterine myocyte to be associated with increased uterine myocyte quiescence and normal gestational length. We hypothesize that ubiquitous uterine gestational stresses experienced in every pregnancy, which have been demonstrated in other systems to activate the UPR, may induce a robust paracrine dissemination of a uterine secretome, for example, glucose-regulated protein 78, with preconditioning-like properties. Furthermore, we speculate that the gestational stress-induced uterine secretome acts to promote both local and systemic tolerance to the ensuing gestational insults, allowing for the maintenance of uterine quiescence. In this context, preterm labor may be the result of a pregnant uterus experiencing a stress it cannot accommodate or when it is unable to host an appropriate UPR resulting in insufficient preconditioning and a diminished local and systemic capacity to tolerate pregnancy-dependent increases in normal gestational stress. This is highly attractive from a clinical viewpoint as we ultimately aim to identify local and systemic adaptations that may serve as preconditioning stimuli for use as a strategy to restore appropriate preconditioning profiles to prolong uterine quiescence in pregnancy.


Subject(s)
Ischemic Preconditioning/methods , Premature Birth/prevention & control , Uterine Contraction , Uterus/physiopathology , Adaptation, Physiological , Animals , Blood Flow Velocity , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/metabolism , Humans , Pregnancy , Premature Birth/metabolism , Premature Birth/physiopathology , Regional Blood Flow , Signal Transduction , Unfolded Protein Response , Uterus/blood supply , Uterus/metabolism
8.
Biol Reprod ; 95(6): 120, 2016 12.
Article in English | MEDLINE | ID: mdl-27733380

ABSTRACT

There is considerable evidence that implicates oxidative stress in the pathophysiology of human pregnancy complications. However, the role and the mechanism of maintaining an antioxidant prosurvival uterine environment during normal pregnancy is largely unresolved. Herein we report that the highly active uterine unfolded protein response plays a key role in promoting antioxidant activity in the uterine myocyte across gestation. The unfolded protein response (UPR) senses the accumulation of misfolded proteins in the endoplasmic reticulum (ER) and activates a signaling network that consists of the transmembrane protein kinase eukaryotic translation initiation factor 2 alpha kinase 3/PKR-like-ER kinase (EIF2AK3), which acts to decrease protein translation levels, allowing for a lowered need for protein folding during periods of ER stress. However, independent of its translational regulatory capacity, EIF2AK3-dependent signals elicit the activation of the transcription factor, nuclear factor erythroid 2-like 2 (NFE2L2) in response to oxidative stress. NFE2L2 binds to antioxidant response elements in the promoters of a variety of antioxidant genes that minimize the opportunities for generation of reactive oxygen intermediates. Our analysis demonstrates that in the absence of EIF2AK3, the uterine myocyte experiences increased levels of reactive oxygen species due to decreased NFE2L2 activation. Elevated levels of intracellular reactive oxygen species were observed in the EIF2AK3 null cells, and this was associated with the onset of apoptotic cell death. These findings confirm the prosurvival and antioxidant role of UPR-mediated EIF2AK3 activation in the context of the human uterine myocyte.


Subject(s)
Endometrium/metabolism , Muscle Cells/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Unfolded Protein Response/physiology , Uterus/metabolism , Animals , Apoptosis/physiology , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Female , Humans , Mice , NF-E2-Related Factor 2/metabolism , Pregnancy , Protein Folding , eIF-2 Kinase/metabolism
9.
Proc Natl Acad Sci U S A ; 112(45): 14090-5, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26504199

ABSTRACT

We previously identified myometrial caspase-3 (CASP3) as a potential regulator of uterine quiescence. We also determined that during pregnancy, the functional activation of uterine CASP3 is likely governed by an integrated endoplasmic reticulum stress response (ERSR) and is consequently limited by an increased unfolded protein response (UPR). The present study examined the functional relevance of uterine UPR-ERSR in maintaining myometrial quiescence and regulating the timing of parturition. In vitro analysis of the human uterine myocyte hTERT-HM cell line revealed that tunicamycin (TM)-induced ERSR modified uterine myocyte contractile responsiveness. Accordingly, alteration of in vivo uterine UPR-ERSR using a pregnant mouse model significantly modified gestational length. We determined that "normal" gestational activation of the ERSR-induced CASP3 and caspase 7 (CASP7) maintains uterine quiescence through previously unidentified proteolytic targeting of the gap junction protein, alpha 1(GJA1); however, surprisingly, TM-induced uterine ERSR triggered an exaggerated UPR that eliminated uterine CASP3 and 7 tocolytic action precociously. These events allowed for a premature increase in myometrial GJA1 levels, elevated contractile responsiveness, and the onset of preterm labor. Importantly, a successful reversal of the magnified ERSR-induced preterm birth phenotype could be achieved by pretreatment with 4-phenylbutrate, a chaperone protein mimic.


Subject(s)
Caspase 3/metabolism , Caspase 7/metabolism , Endoplasmic Reticulum Stress/physiology , Myometrium/physiology , Pregnancy/physiology , Unfolded Protein Response/physiology , Uterus/metabolism , Analysis of Variance , Animals , Cell Line , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , Phenylbutyrates/pharmacology , Pregnancy/drug effects , Progesterone/blood , Reverse Transcriptase Polymerase Chain Reaction
10.
J Clin Invest ; 125(7): 2808-24, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26098214

ABSTRACT

The precise mechanisms that lead to parturition are incompletely defined. Surfactant protein-A (SP-A), which is secreted by fetal lungs into amniotic fluid (AF) near term, likely provides a signal for parturition; however, SP-A-deficient mice have only a relatively modest delay (~12 hours) in parturition, suggesting additional factors. Here, we evaluated the contribution of steroid receptor coactivators 1 and 2 (SRC-1 and SRC-2), which upregulate SP-A transcription, to the parturition process. As mice lacking both SRC-1 and SRC-2 die at birth due to respiratory distress, we crossed double-heterozygous males and females. Parturition was severely delayed (~38 hours) in heterozygous dams harboring SRC-1/-2-deficient embryos. These mothers exhibited decreased myometrial NF-κB activation, PGF2α, and expression of contraction-associated genes; impaired luteolysis; and elevated circulating progesterone. These manifestations also occurred in WT females bearing SRC-1/-2 double-deficient embryos, indicating that a fetal-specific defect delayed labor. SP-A, as well as the enzyme lysophosphatidylcholine acyltransferase-1 (LPCAT1), required for synthesis of surfactant dipalmitoylphosphatidylcholine, and the proinflammatory glycerophospholipid platelet-activating factor (PAF) were markedly reduced in SRC-1/-2-deficient fetal lungs near term. Injection of PAF or SP-A into AF at 17.5 days post coitum enhanced uterine NF-κB activation and contractile gene expression, promoted luteolysis, and rescued delayed parturition in SRC-1/-2-deficient embryo-bearing dams. These findings reveal that fetal lungs produce signals to initiate labor when mature and that SRC-1/-2-dependent production of SP-A and PAF is crucial for this process.


Subject(s)
Maternal-Fetal Exchange/physiology , Nuclear Receptor Coactivator 1/physiology , Nuclear Receptor Coactivator 2/physiology , Parturition/physiology , 1-Acylglycerophosphocholine O-Acyltransferase/deficiency , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Female , Fetal Organ Maturity , Heterozygote , Lung/embryology , Lung/physiology , Luteolysis , Male , Maternal-Fetal Exchange/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Nuclear Receptor Coactivator 1/deficiency , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 2/deficiency , Nuclear Receptor Coactivator 2/genetics , Platelet Activating Factor/deficiency , Pregnancy , Promoter Regions, Genetic , Pulmonary Surfactant-Associated Protein A/deficiency , Signal Transduction , Transcriptional Activation , Uterus/physiology
11.
Endocrinology ; 154(12): 4873-84, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24140717

ABSTRACT

A successful postpartum involution permits the postnatal uterus to rapidly regain its prepregnancy function and size to ultimately facilitate an ensuing blastocyst implantation. This study investigates the molecular mechanisms that govern the initiation of the involution process by examining the signaling events that occur as the uterus transitions from the pregnant to postnatal state. Using mouse and baboon uteri, we found a remarkable cross-species conservation at the signal transduction level as the pregnant uterus initiates and progresses through the involution process. This study originated with the observation of elevated levels of caspase-3 activation in both the laboring mouse and baboon uterus, which we found to be apoptotic in nature as evidenced by the concurrent appearance of cleaved poly(ADP-ribose) polymerase. We previously defined a nonapoptotic and potential tocolytic role for uterine caspase-3 during pregnancy regulated by increased antiapoptotic signaling mediated by myeloid cell leukemia sequence 1 and X-linked inhibitor of apoptosis. In contrast, this study determined that diminished antiapoptotic signaling in the postpartum uterus allowed for both endometrial apoptotic and myometrial autophagic episodes, which we speculate are responsible for the rapid reduction in size of the postpartum uterus. Using our human telomerase immortalized myometrial cell line and the Simian virus-40 immortalized endometrial cell line (12Z), we demonstrated that the withdrawal of antiapoptotic signaling was also an upstream event for both the autophagic and apoptotic processes in the human uterine myocyte and endometrial epithelial cell.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Autophagy , Caspase 3 , Cell Line , Female , In Situ Nick-End Labeling , Labor, Obstetric/physiology , Mice , Microtubule-Associated Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myometrium/cytology , Papio anubis , Postpartum Period , Pregnancy , Signal Transduction , Up-Regulation , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
12.
PLoS One ; 8(9): e75152, 2013.
Article in English | MEDLINE | ID: mdl-24058658

ABSTRACT

We have previously proposed that uterine caspase-3 may modulate uterine contractility in a gestationally regulated fashion. The objective of this study was to determine the mechanism by which uterine caspase-3 is activated and consequently controlled in the pregnant uterus across gestation. Utilizing the mouse uterus as our gestational model we examined the intrinsic and extrinsic apoptotic signaling pathways and the endoplasmic reticulum stress response as potential activators of uterine caspase-3 at the transcriptional and translational level. Our study revealed robust activation of the uterine myocyte endoplasmic reticulum stress response and its adaptive unfolded protein response during pregnancy coinciding respectively with increased uterine caspase-3 activity and its withdrawal to term. In contrast the intrinsic and extrinsic apoptotic signaling pathways remained inactive across gestation. We speculate that physiological stimuli experienced by the pregnant uterus likely potentiates the uterine myocyte endoplasmic reticulum stress response resulting in elevated caspase-3 activation, which is isolated to the pregnant mouse myometrium. However as term approaches, activation of an elevated adaptive unfolded protein response acts to limit the endoplasmic reticulum stress response inhibiting caspase-3 resulting in its decline towards term. We speculate that these events have the capacity to regulate gestational length in a caspase-3 dependent manner.


Subject(s)
Caspase 3/metabolism , Endoplasmic Reticulum Stress/physiology , Muscle Cells/enzymology , Myometrium/enzymology , Pregnancy/physiology , Signal Transduction/physiology , Animals , Enzyme Activation/physiology , Female , Mice , Muscle Cells/cytology , Myometrium/cytology , Protein Biosynthesis/physiology , Transcription, Genetic/physiology
13.
Proc Natl Acad Sci U S A ; 109(19): 7529-34, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22529366

ABSTRACT

During pregnancy, uterine quiescence is maintained by increased progesterone receptor (PR) activity, but labor is facilitated by a series of events that impair PR function. Previously, we discovered that miR-200 family members serve as progesterone (P(4))-modulated activators of contraction-associated genes in the pregnant uterus. In this study, we identified a unique role for miR-200a to enhance the local metabolism of P(4) in myometrium and, thus, decrease PR function during the progression toward labor. miR-200a exerts this action by direct repression of STAT5b, a transcriptional repressor of the P(4)-metabolizing enzyme 20α-hydroxysteroid dehydrogenase (20α-HSD). We observed that miR-200a expression increased and STAT5b expression coordinately decreased in myometrium of mice as they progressed to labor and in laboring myometrium from pregnant women. These changes were associated with a dramatic increase in expression and activity of 20α-HSD in laboring myometrium from mouse and human. Notably, overexpression of miR-200a in cultured human myometrial cells (hTERT-HM) suppressed STAT5b and increased 20α-HSD mRNA levels. In uterine tissues of ovariectomized mice injected with P(4), miR-200 expression was significantly decreased, STAT5b expression was up-regulated, and 20α-HSD mRNA was decreased, but in 15 d postcoitum pregnant mice injected with the PR antagonist RU486, preterm labor was associated with increased miR-200a, decreased STAT5b, and enhanced 20α-HSD expression. Taken together, these findings implicate miR-200a as an important regulator of increased local P(4) metabolism in the pregnant uterus near term and provide insight into the importance of miR-200s in the decline in PR function leading to labor.


Subject(s)
Labor, Obstetric/genetics , MicroRNAs/genetics , Obstetric Labor, Premature/genetics , Receptors, Progesterone/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , Animals , Cells, Cultured , Female , Gene Expression/drug effects , Hormone Antagonists/pharmacology , Humans , Immunoblotting , Labor, Obstetric/metabolism , Male , Mice , Mice, Inbred ICR , Mifepristone/pharmacology , Myometrium/cytology , Myometrium/metabolism , Obstetric Labor, Premature/metabolism , Ovariectomy , Pregnancy , Progesterone/metabolism , Progesterone/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Uterus/drug effects , Uterus/metabolism
14.
Mol Endocrinol ; 26(2): 320-30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22194343

ABSTRACT

Our group has previously identified elevated levels of nonapoptotic active caspase 3 (CASP3) accompanied by increased prosurvival, antiapoptotic signaling in the pregnant mouse uterus during late gestation. We speculated that increased antiapoptotic signaling desensitized the pregnant uterine myocyte to the apoptotic action of uterine CASP3. This current study examines the mechanism by which the pregnant myocyte gains resistance to the apoptotic effects of increased uterine CASP3. Using both primary human pregnant fundal myometrial cultures and the telomerase-immortalized human uterine myocyte cell line (hTERT) as our model systems, uterine myocytes were exposed to UV irradiation and Fas ligand to stimulate both the intrinsic and extrinsic apoptotic pathways. Stimulation of either the intrinsic or extrinsic apoptotic pathways resulted in elevated levels of uterine myocyte CASP3. However, apoptotic cell death was restricted to CASP3 activated by intrinsic stimulation via UV light. In contrast Fas ligand-mediated CASP3 activation was accompanied by increased antiapoptotic signaling mimicking our in vivo observations in the pregnant mouse uterus. Using small interfering RNA to inhibit antiapoptotic signaling, we determined the ability of the human uterine myocyte to resist apoptotic cell death in the absence of the prosurvival, antiapoptotic signaling. Accordingly, suppression of antiapoptotic signaling specifically mediated by myeloid cell leukemia sequence 1 was sufficient to sensitize the uterine myocyte to undergo apoptotic cell death. These data demonstrate that elevated myeloid cell leukemia sequence 1 levels are sufficient to confer apoptotic resistance on the human uterine myocyte despite highly elevated levels of active CASP3.


Subject(s)
Apoptosis , Caspase 3/physiology , Muscle Cells/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Uterus/cytology , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Catalase/genetics , Catalase/metabolism , Cell Line , DNA Fragmentation , Enzyme Activation , Fas Ligand Protein/pharmacology , Female , Gene Expression Regulation , Humans , Muscle Cells/enzymology , Muscle Cells/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Pregnancy , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , Ultraviolet Rays
15.
Biol Reprod ; 85(2): 417-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21566000

ABSTRACT

Preserving the uterus in a state of relative quiescence is vital to the maintenance of a successful pregnancy. Elevated cytoplasmic levels of uterine caspase 3 during pregnancy have been proposed as a potential regulator of uterine quiescence through direct targeting and disabling of the uterine contractile architecture. However, despite highly elevated levels of uterine caspase 3 during pregnancy, there is minimal evidence of apoptosis. This current study defines the mechanism whereby the pregnant uterine myocyte may harness the tocolytic activity of active caspases while avoiding apoptotic cell death. Using the pregnant mouse model, we have analyzed the uterus for changes in pro- and antiapoptotic signaling patterns associated with the advancing stages of pregnancy. Briefly, we have found that members of the IAP family, such as SURVIVIN and XIAP, and the Bcl2 family members, such as MCL1, are elevated in the uterine myocyte during late gestation. The IAP family members are the only endogenous inhibitors of active caspase 3, and MCL1 limits activation of caspase 3 by suppressing proapoptotic signaling. Elevated XIAP levels partner with SURVIVIN, resulting in increased levels of the antiapoptotic MCL1 via NFKB activation; these together have the potential to limit both the activity and level of active caspase 3 in the pregnant uterus as term approaches. We propose that modification of these antiapoptotic signaling partners allows the pregnant uterus to escape the apoptotic action of elevated active caspase 3 levels but also functions to limit the levels of active uterine caspase 3 near term.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , NF-kappa B/metabolism , Uterus/physiology , Animals , Caspase 3/genetics , DNA/metabolism , Female , Genome , Mice , NF-kappa B/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Pregnancy , Uterus/cytology
16.
Biol Reprod ; 80(5): 928-34, 2009 May.
Article in English | MEDLINE | ID: mdl-19144964

ABSTRACT

The appropriate timing of the onset of labor is critical to a successful pregnancy, with potentially devastating consequences resulting to both the mother and child with the onset of preterm labor. In this study, we tested the central hypothesis that progesterone maintains uterine quiescence through regulation of active uterine caspase 3. Using the mouse as our model system, we examined, by Western blot analysis, levels of active caspase 3 and its association with the degradation of uterine contractile proteins during pregnancy. Our data demonstrate that caspase 3-specific cleavage fragments of uterine myocyte contractile proteins are elevated in late gestation. Prior to the onset of labor, active caspase 3 levels and fragmentation of the uterine myocyte contractile proteins decline. We postulate that uterine caspase 3 acts as an anticontractile agent maintaining uterine quiescence through degradation of uterine contractile proteins during late pregnancy. We propose that decreased progesterone action during the final days of pregnancy controls the timing of the onset of uterine contractions by removing the anticontractile action of the apoptotic protein caspase 3 locally in the pregnant myometrium.


Subject(s)
Caspase 3/metabolism , Progesterone/metabolism , Uterus/metabolism , Actins/genetics , Actins/metabolism , Animals , Base Sequence , Caspase 3/genetics , Female , Humans , Labor Onset/metabolism , Mice , Mice, Inbred ICR , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Pregnancy , Progesterone/pharmacology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uterine Contraction/metabolism , Uterus/drug effects
17.
Mol Endocrinol ; 20(4): 764-75, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16339279

ABSTRACT

Progesterone acting via the progesterone receptor (PR) plays a critical role in maintaining uterine quiescence during pregnancy. In the present study, we tested the hypothesis that the transactivating capability of the PR is down-regulated in the myometrium at term by a change in uterine PR isoform ratio resulting from local activation of the nuclear factor (NF)-kappaB pathway. Overexpression of the truncated PR-C isoform in human myometrial cells inhibited PR-B transactivation. Expression of PR isoforms, PR-A, PR-B, and PR-C, was characterized by immunoblotting and quantitative PCR (Q-PCR) in fundal and lower uterine segment myometrium from pregnant women in labor and not in labor and in the pregnant mouse uterus during late gestation. We observed a marked increase in levels of PR-C and transcriptionally active PR-B specifically in fundal myometrium of women in labor. In pregnant mouse uterus, levels of PR-B and PR-C also increased between 15 days post coitum and term, whereas expression of PR-A was dramatically up-regulated at 19 days post coitum. In studies of uterine tissues of mice injected intraamniotically with surfactant protein A and of human myometrial and T47D breast cancer cells in culture, up-regulation of PR isoform expression was observed in response to activation of the NF-kappaB pathway. Chromatin immunoprecipitation analysis revealed IL-1beta induced binding of NF-kappaB to the PR promoter. Collectively, these findings suggest that up-regulation of inhibitory PR isoform expression by NF-kappaB activation in both laboring human fundus and pregnant mouse uterus near term may inhibit PR transactivation and thereby lead to a loss of uterine quiescence and the onset of labor.


Subject(s)
Labor, Obstetric/genetics , Labor, Obstetric/metabolism , Myometrium/metabolism , NF-kappa B/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , Female , Humans , In Vitro Techniques , Labor Onset/genetics , Labor Onset/metabolism , Mice , Mice, Inbred ICR , Pregnancy , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Progesterone/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcriptional Activation , Transfection , Up-Regulation
18.
J Steroid Biochem Mol Biol ; 93(2-5): 113-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15860253

ABSTRACT

The signals that lead to the initiation of parturition have remained a mystery. We postulate that in humans and other mammals, uterine quiescence is maintained by increased progesterone receptor (PR) transcriptional activity, and spontaneous labor is initiated or facilitated by a concerted series of biochemical events that negatively impact PR function. In recent studies, we have obtained compelling evidence to suggest that the fetus signals the initiation of labor by secretion into amniotic fluid of major lung surfactant protein, SP-A. SP-A expression is developmentally regulated in fetal lung and is secreted into amniotic fluid in high concentrations near term (after 17 days postcoitum [dpc] in the mouse). We found that injection of exogenous SP-A into mouse amniotic fluid at 15 dpc caused preterm labor. SP-A activated amniotic fluid macrophages in vitro to express nuclear factor kappaB (NF-kappaB) and interleukin-1beta (IL-1beta). These macrophages, which are of fetal origin, migrate to the pregnant uterus causing an inflammatory response and increased uterine NF-kappaB activity. We suggest that the increase in NF-kappaB within the maternal uterus both directly increases expression of genes that promote uterine contractility and negatively impacts the capacity of the PR to maintain uterine quiescence, contributing to the onset of labor. Our findings, therefore, indicate that SP-A secreted into amniotic fluid by the maturing fetal lung serves as a hormone of parturition.


Subject(s)
Parturition/physiology , Amniotic Fluid/metabolism , Animals , Female , Fetus/physiology , Humans , Inflammation/physiopathology , Interleukin-1/metabolism , Lung/physiology , Mice , Models, Biological , NF-kappa B/metabolism , Obstetric Labor, Premature/etiology , Obstetric Labor, Premature/physiopathology , Pregnancy , Pulmonary Surfactant-Associated Protein A/physiology , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 101(14): 4978-83, 2004 Apr 06.
Article in English | MEDLINE | ID: mdl-15044702

ABSTRACT

Parturition is timed to begin only after the developing embryo is sufficiently mature to survive outside the womb. It has been postulated that the signal for the initiation of parturition arises from the fetus although the nature and source of this signal remain obscure. Herein, we provide evidence that this signal originates from the maturing fetal lung. In the mouse, secretion of the major lung surfactant protein, surfactant protein A (SP-A), was first detected in amniotic fluid (AF) at 17 days postcoitum, rising progressively to term (19 days postcoitum). Expression of IL-1beta in AF macrophages and activation of NF-kappaB in the maternal uterus increased with the gestational increase in SP-A. SP-A stimulated IL-1beta and NF-kappaB expression in cultured AF macrophages. Studies using Rosa 26 Lac-Z (B6;129S-Gt(rosa)26Sor) (Lac-Z) mice revealed that fetal AF macrophages migrate to the uterus with the gestational increase in AF SP-A. Intraamniotic (i.a.) injection of SP-A caused preterm delivery of fetuses within 6-24 h. By contrast, injection of an SP-A antibody or NF-kappaB inhibitor into AF delayed labor by >24 h. We propose that augmented production of SP-A by the fetal lung near term causes activation and migration of fetal AF macrophages to the maternal uterus, where increased production of IL-1beta activates NF-kappaB, leading to labor. We have revealed a response pathway that ties augmented surfactant production by the maturing fetal lung to the initiation of labor. We suggest that SP-A secreted by the fetal lung serves as a hormone of parturition.


Subject(s)
Lung/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Uterine Contraction/physiology , Animals , Base Sequence , DNA Primers , Female , Immunohistochemistry , Interleukin-1/genetics , Lung/embryology , Macrophages/metabolism , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Uterus/cytology , Uterus/metabolism , Uterus/physiology
20.
Proc Natl Acad Sci U S A ; 100(16): 9518-23, 2003 Aug 05.
Article in English | MEDLINE | ID: mdl-12886011

ABSTRACT

The molecular events that lead to the onset of labor in humans and in other mammalian species remain unclear. We propose that a decline in coactivators containing histone acetylase activity in myometrium may contribute to the onset of labor by impairing the function of the progesterone-progesterone receptor (PR) complex. As assessed by semiquantitative and real-time RT-PCR, immunohistochemistry, and immunoblotting, expression of the PR coactivators cAMP-response element-binding protein (CREB)-binding protein and steroid receptor coactivators 2 and 3 was decreased in fundal uterine tissue of women in labor. Using the mouse as an animal model, we also found decreased coactivator levels in uterine tissues at term. In both human and mouse, the levels of acetylated histone H3 were also decreased in uterine tissues at term. Administration of trichostatin A, a specific and potent histone deacetylase inhibitor, to pregnant mice late in gestation increased histone acetylation and delayed the initiation of parturition by 24-48 h, suggesting the functional importance of the decline in histone acetylation in the initiation of labor. These findings suggest that the decline in PR coactivator expression and in histone acetylation in the uterus near term may impair PR function by causing a functional progesterone withdrawal. The resulting decrease in expression of PR-responsive genes should increase sensitivity of the uterus to contractile stimuli.


Subject(s)
Receptors, Progesterone/metabolism , Uterus/metabolism , Acetyltransferases , Animals , CREB-Binding Protein , Cell Nucleus/metabolism , DNA Primers/metabolism , Enzyme Inhibitors/pharmacology , Female , Histone Acetyltransferases , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Immunoblotting , Immunohistochemistry , Labor, Obstetric , Mice , Myometrium/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2 , Nuclear Receptor Coactivator 3 , Oncogene Proteins , Pregnancy , Progesterone/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/biosynthesis , Trans-Activators/metabolism , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...